Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

ARTICLE

https://doi.org/10.1038/s41467-021-22507-3 OPEN

Extracellular cap domain is an essential component


of the TRPV1 gating mechanism
Kirill D. Nadezhdin1,5, Arthur Neuberger1,5, Yury A. Nikolaev2, Lyle A. Murphy2, Elena O. Gracheva2,3,4,
Sviatoslav N. Bagriantsev 2 & Alexander I. Sobolevsky 1 ✉
1234567890():,;

Transient receptor potential (TRP) channels are polymodal molecular sensors involved in
numerous physiological processes and implicated in a variety of human diseases. Several
structures of the founding member of the TRP channel family, TRPV1, are available, all of
which were determined for the protein missing the N- and C-termini and the extracellular S5-
P-loop. Here, we present structures of the full-length thirteen-lined ground squirrel
TRPV1 solved by cryo-EM. Our structures resolve the extracellular cap domain formed by the
S5-P-loops and the C-terminus that wraps around the three-stranded β-sheet connecting
elements of the TRPV1 intracellular skirt. The cap domain forms a dome above the pore’s
extracellular entrance, with four portals leading to the ion conductance pathway. Deletion of
the cap increases the TRPV1 average conductance, reduces the open probability and affects
ion selectivity. Our data show that both the termini and the cap domain are critical deter-
minants of TRPV1 function.

1 Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA. 2 Department of Cellular and Molecular Physiology, Yale

University School of Medicine, New Haven, CT, USA. 3 Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA. 4 Program in
Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA. 5These authors contributed equally: Kirill D.
Nadezhdin, Arthur Neuberger. ✉email: as4005@cumc.columbia.edu

NATURE COMMUNICATIONS | (2021)12:2154 | https://doi.org/10.1038/s41467-021-22507-3 | www.nature.com/naturecommunications 1


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-021-22507-3

I
n mammals, the detection of high temperature is carried out domain that shapes portals for ions to enter the central channel
by terminal endings of primary afferent neurons expressing pore and is critical for channel conductance, open probability
thermosensitive ion channels of the transient receptor poten- and ion selectivity. We also show that the C-terminus of the full-
tial (TRP) family1. Several mammalian species have evolved the length TRPV1 wraps around the three-stranded β-sheet making
ability to comfortably withstand high environmental heat for a numerous molecular interactions that strengthen intersubunit
long period of time, which allowed them to inhabit otherwise interfaces and stabilize the intracellular skirt.
prohibitively hot ecological niches. Among these are small diurnal
rodents, including 13-lined ground squirrels (Ictidomys tride-
cemlineatus), who developed tolerance to environmental heat via Results and discussion
modification of the primary noxious heat sensor, TRPV1 Apo-state structure. We expressed full-length wild-type
(refs. 2–4). In contrast to TRPV1 orthologues from rat (rTRPV1) sqTRPV1 (ref. 5) in HEK 293 cells and subjected the purified apo
and other mammals, ground squirrel TRPV1 (sqTRPV1) fails to protein to cryo-EM (Supplementary Fig. 1 and Supplementary
activate by heating in a physiologically relevant temperature Table 1). The resulting structure (Fig. 1a–c) has a similar overall
range, even though the channel is sensitive to chemical agonists, shape and architecture, as the previously published structures of
such as acid and capsaicin5. The available structures of rTRPV1 truncated rTRPV1 (refs. 6–8; Supplementary Fig. 2). However, of
revealed the overall architecture and key elements of the gating the three regions missing in rTRPV1—the N-terminus, the S5-P-
machinery6–8, but all of these structures were solved for a trun- loop and the C-terminus—the latter two appear to fold into
cated construct that lacked the N- and C-termini, as well as the structural elements in the context of full-length sqTRPV1. The C-
extracellular S5-P-loop. In fact, these missing regions have been terminus of sqTRPV1 represents an extended polypeptide that
implicated in the mechanism of temperature sensitivity9–18, wraps around the three-stranded β-sheet (Fig. 1d), which con-
which remains poorly understood. nects the ankyrin repeat domains of the neighbouring subunits to
In this work, we study the full-length sqTRPV1 using cryo- form the typical for TRPV channels intracellular skirt. Electro-
electron microscopy (cryo-EM) combined with electrophysiology static and hydrophobic interactions between the C-terminus, the
and mutagenesis to understand the general mechanisms of TRPV1 three-stranded β-sheet and the C-terminal hook of one subunit,
function. We show that the S5-P-loops form an extracellular cap and the loops of ankyrin repeats AR2–5 of the neighbouring

a b
Cap

Out B
90° C
A C

In

C-terminus

B D A
D

90°

c d pre-S1
A AR5

E3 D
E2
D
E1

AR4
B
AR3

Hook
C AR2 N126
A AR1

Fig. 1 Structure of full-length sqTRPV1. a–c Side (a), top (b) and bottom (c) views of full-length sqTRPV1 structure with four subunits coloured differently,
the cap domain in blue and the C-terminus in pink. Dark blue sphere is a putative chloride ion. d Close-up view of the C-terminus with the residues
contributing to interactions with other domains shown in sticks. Asparagine N126, the serine substitution of which converts weakly temperature-sensitive
sqTRPV1 into highly temperature-sensitive rTRPV1-like channel, is labelled.

2 NATURE COMMUNICATIONS | (2021)12:2154 | https://doi.org/10.1038/s41467-021-22507-3 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-021-22507-3 ARTICLE

subunit make the intersubunit interfaces stronger. As a result, the To test the importance of the cap domain for the TRPV1
intracellular skirt of full-length sqTRPV1 is less dynamic than function, we generated a mutant channel lacking residues
that of truncated rTRPV1. Indeed, the entire skirt is well-resolved 606–628 (sqTRPV1 d606–628, Supplementary Fig. 3). Electro-
in the cryo-EM density of full-length sqTRPV1, while density physiological recordings in Xenopus oocytes showed that
for two peripheral of the six ankyrin repeats, as well as the sqTRPV1 d606–628 lacks heat activation (Supplementary Fig. 4a),
C-terminal hook domain preceding the third strand of the three- similar to wild-type channel5. The amplitude histograms of cell-
stranded β-sheet in truncated rTRPV1 was missing6–8. Other attached single-channel recordings from HEK293T cells revealed
than these differences, apo-state structures of sqTRPV1 and that deletion of the cap domain produced a significant increase in
rTRPV1 are quite similar (Supplementary Fig. 2a, d), suggesting single-channel conductance from 125 ± 4.9 pS for wild type to
that whenever the complete termini are present in rTRPV1, they 174.6 ± 6.6 pS for the d606–628 mutant (P = 0.003, t test,
are likely to interact with the rest of the protein in the same way Fig. 2c–e)26. Compared to wild-type sqTRPV1, the d606–628
as in sqTRPV1. mutant exhibited flickering behaviour, in part due to the
Similar to sqTRPV1, the extended C-terminus that wraps significantly decreased mean open dwell time (Fig. 2f and
around the three-stranded β-sheet has also been observed in cryo- Supplementary Fig. 4b). In addition, we expressed sqTRPV1 wt
EM structures of other temperature-sensitive TRPV channels and sqTRPV1 d606–628 in Xenopus oocytes, and compared their
TRPV2–3 (refs. 19–22). Compared to TRPV2–3, the C-terminus in reversal potential, Erev, by recording currents in response to
the full-length sqTRPV1, after wrapping around the β-sheet, different concentrations of capsaicin. At both 200 nM and 2 µM
makes a U-turn and follows the edge of the skirt, providing of capsaicin, sqTRPV1 wt and sqTRPV1 d606–628 showed
additional interdomain interactions (Fig. 1d). Unwrapping of the significantly different values of Erev, suggesting that the deletion
C-terminus and binding of an otherwise unfolded region of the N- of the S5-P-loop affects ion selectivity of sqTRPV1 (Supplemen-
terminus have been recently shown to accompany temperature- tary Fig. 4c). Together, our functional data show that the cap
induced opening of TRPV3 (ref. 23). Similarly, both N- and C- domain is an essential component of the TRPV1 gating
termini may contribute to temperature gating of TRPV1. mechanism.
Supporting this idea, the serine substitution of asparagine N126
converted weakly temperature-sensitive sqTRPV1 into highly
Lipids and ligands. The apo-state structure of the full-length
temperature-sensitive rTRPV1-like channel5. N126 is located at
sqTRPV1 shows at least nine well-resolved densities of annular
the end of the AR1 helix that is in direct contact with AR2 helix
lipids per channel subunit (Fig. 3a, b). The majority of densities
contacting the C-terminus (Fig. 1d). Thus, alteration of the
have a clear head-and-two-tails appearance and were modelled
AR1–AR2 interaction is expected to have an immediate allosteric
with phosphatidylcholine. The exception is the lipid at site 1 or
effect on the binding of the C-terminus. In contrast, in
the vanilloid binding site, which in structures of rTRPV1
temperature-insensitive TRPV5–6, the C-terminus does not wrap
accommodates agonists resiniferatoxin (RTX) and capsaicin, and
around the three-stranded β-sheet, and the N-terminal helix,
antagonist capsazepine6–8. Similar to the apo-state structure of
which pillars the corners of the intracellular skirt24,25, plays the
rTRPV1 (ref. 7), the vanilloid site in the apo-state structure of
role of an intersubunit “glue”.
sqTRPV1 contains density that is best fit by phosphatidylinositol
(PI, Fig. 3c). The head of PI is accommodated by polar and
charged residues of the linker domain (R411 and H412), the
Extracellular cap. The S5-P-loops of the full-length sqTRPV1
S2–S3 loop (D511 and Y513), the S4–S5 linker (R559, E572 and
assemble into an extracellular cap domain that forms a dome
K573) and the TRP helix (Q702), while the PI tails face hydro-
above the pore extracellular entry (Fig. 1a, b). The cryo-EM
phobic residues of S3 and S4 from one subunit, and S5 and S6
density for the cap is relatively weak and is completely missing for
from the neighbouring subunit.
residues 608–617. This can be explained by the dynamic nature
To get an insight into ligand binding to full-length TRPV1, we
of the S5-P-loops and by possible spontaneous crosslinking of
determined structures of sqTRPV1 in complex with agonists RTX
cysteines C623, which in the fourfold symmetrical model of
and capsaicin, which both potently activate sqTRPV1, similar to
sqTRPV1 come close together at the centre of the cap with 5.5 Å
the rat orthologue (Supplementary Fig. 4d)5,27. In agreement with
distances between their Cα atoms. In case such crosslinking does
the previous structures of rTRPV1, both RTX and capsaicin bind
happen, the population of channels that contribute to cryo-EM
to the vanilloid site and are coordinated by residues in S2–S3 loop,
reconstruction is likely heterogeneous and includes particles with
S3, S4 and S4–S5 linker of one subunit, and S5 and S6 of the
non-symmetrical (one cysteine crosslink), twofold symme-
neighbouring subunit (Fig. 3d, e and Supplementary Fig. 5). In the
trical (two crosslinks) and fourfold symmetrical (no crosslinks)
apo state, the same residues contribute to the binding of PI, which
cap domains.
resides deeper in the vanilloid site towards the linker domain and
Strong density in the middle of the cap, which we interpreted
pushes away the side chain of tyrosine Y513. Binding of both RTX
as a chloride ion coordinated by arginines R624, represents a
and capsaicin results in shortening the distance between the
likely barrier for cation conductance along the central axis of the
hydrogen bond-forming sidechains of T552 and E572, and
channel. On the other hand, four side portals in the dome lead to
movement of the S4–S5 linker away from the pore (Supplemen-
the ion-conducting pore extracellular entrance (Fig. 2a, b). Each
tary Fig. 5b, c). This S4–S5 linker movement was proposed to be
portal is formed by the N-terminal part of the S5-P-loop of one
coupled to S6 movement and pore opening28. While binding of PI,
subunit and the C-terminal part of the S5-P-loop of the
RTX and capsaicin to full-length sqTRPV1 is nearly identical to
neighbouring subunit, and contains numerous negatively charged
their binding to truncated rTRPV1, there is a notable difference in
residues. Correspondingly, the overall negative charge of each
the resulting architecture of the ion channel pore.
individual portal (Fig. 2b) creates a favourable environment for
passage of cations, and suggests a possible role of the cap domain
in TRPV1 conductance and ion selectivity. The missing residues Ion channel pore. The pore of the full-length sqTRPV1 in the
608–617 can potentially form a loop capable of occluding the side apo and RTX-bound states (Fig. 4a, b) resembles the pore of the
portals, thus providing an additional way to regulate the ion truncated rTRPV1 in the apo state6–8 (Supplementary Fig. 2).
channel conductance. Measurements of the pore radius confirm that both the selectivity

NATURE COMMUNICATIONS | (2021)12:2154 | https://doi.org/10.1038/s41467-021-22507-3 | www.nature.com/naturecommunications 3


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-021-22507-3

a R624 b Side portal

K605 R619
R619 K605
D603
E653 D603
D627 E650
D648 E650 D648 E602
E602 E638
S5 S6
S1 K641
P
P S4
S2 S3
S3
S5 S6
S4 S1 S2

c
sqTRPV1 wt sqTRPV1 d606-628
10 pA
50 ms

mV: mV:

+90 +80

+80 +70

+70 +60

+60 +50

10 pA
d 500 ms 20
20
Count (N x 100)
Count (N x 100)

10
10

0 0
0 10 20 30 0 10 20 30
Amplitude (pA) Amplitude (pA)
e f 8
20
1st level mean open time (ms)

sqTRPV1 wt
sqTRPV1 d606-628
**
15 6
Amplitude (pA)

10 4
174 pS

5 2

125 pS
0 0
0 50 100 150 wt d606-628
Voltage (mV)

filter and the lower gate of the channel are completely closed for capture the open-state structure of sqTRPV1, which can be
permeation of water and ions (Fig. 4d). Interestingly, the presence opened functionally without help of DKTX (Supplementary
of DKTX bound to rTRPV1 together with RTX leads to sig- Fig. 4d).
nificant pore widening and opening of the channel6–8 (Supple- The narrow constriction at the lower gate of sqTRPV1 in the
mentary Fig. 2e). It remains to be determined whether the open- apo and RTX-bound states is formed by the S6 residue I681,
state structure of rTRPV1 can be determined in the absence which is homologous to I679 that determines the narrowest
of DKTX. However, binding of RTX alone was not enough to diameter of the lower pore in all published structures of rTRPV1

4 NATURE COMMUNICATIONS | (2021)12:2154 | https://doi.org/10.1038/s41467-021-22507-3 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-021-22507-3 ARTICLE

Fig. 2 Cap domain and its role in TRPV1 function. a Ribbon diagram of the sqTRPV1 extracellular portion with subunits in different colour and the cap
domain in blue. Charged residues are shown as sticks. b Surface view of the same region coloured by electrostatic potential with positively charged areas in
blue, negatively charged in red and neutral in white. c Representative traces from single-channel recording of spontaneous openings of wild-type sqTRPV1
and d606–628 mutant at different voltages in cell-attached patch from HEK293T cells at room temperature. Black arrows indicate the position of the
baseline current. Upward single-channel deflections represent outward current. d Amplitude histograms corresponding to the channel activity at 90 mV for
wild-type sqTRPV1 and d606–628 mutant. e Current–voltage relationship for wild-type sqTRPV1 and d606–628 mutant with its single-channel
conductances, fitted to the linear equation (wt, N = 5 cells; d606–628, N = 9 cells). Source data are provided as a Source data file. f Mean open dwell time
of wild-type sqTRPV1 and d606–628 mutant channels (at 70 mV). Each dot represents individual cell (N = 5 cells for each construct). Data are mean ±
SEM. **P = 0.0074, two-sided t test).

a Out
b 2 4
5
3
6 6

In
C
9

D 1
B 9
8

A
7

c L664 d S5 L664 e L664


S1 S4
S4 S4 F545
S5 F545 F545 A667
A667 F593 A667 S2 S1 S3 A548
S2 A548 S2 A548 M549
M549 M549 S5 F593 S6
F593 S6 RTX F589
S3 F589 N553 N553 T552
T552 T552
L671 L671 S6 L671
N553 F589 Cap
L517 PI L517 I575
L517
M516
M516 M516 Y513 I575
I575 L576 L576
Y513 S3 Y513
L576
E572
S5
S5 R559
R559
S1 E572 S5
E572
I705 K573 I705 R559 K573
S6 I705 D511 K573
Q702 D511 I698 Q702 I698
D511 S6 Q702
I698

L701
L701 L701 H412
H412 H412 R411 TRP helix
R411 TRP helix
R411 TRP helix

Fig. 3 Lipids and ligands. a Cryo-EM density for the apo-state full-length sqTRPV1, with the lipid densities coloured red or blue. b Close-up view of the
membrane region, with the molecules of lipid shown in sticks and numbered. c–e Close-up view of the vanilloid site (site 1 in b) in the apo-state structure
harbouring the molecule of phosphatidylinositol (c), RTX-bound (d) and capsaicin-bound (e) structures. The molecules of phosphatidylinositol, RTX and
capsaicin are shown as yellow, blue and green sticks, respectively, with the corresponding cryo-EM density shown in red.

(refs. 6–8). In contrast, the narrow constriction of the lower pore S6 can happen in TRPV1 and propose that this transition is an
in the capsaicin-bound structure of sqTRPV1 is formed by the S6 essential feature of TRPV1 gating.
residue M684, while I681 faces away from the pore (Fig. 4c). This Measurements of the pore radius suggest that the ion channel
change occurs because S6, which contains a central π-bulge in of sqTRPV1 in the capsaicin-bound state is in a closed non-
other conformations of sqTRPV1 and rTRPV1, becomes entirely conducting conformation (Fig. 4d). The channel is too narrow to
α-helical in the capsaicin-bound structure of sqTRPV1. While the conduct water or ions at both the selectivity filter and the lower
α-helical S6 has not been observed in TRPV1 structures before6–8, pore, where the narrow constriction is formed by M684. In
it has been predicted29 and has also been observed in structures of contrast, despite remaining non-conducting, the lower pore in the
other representatives of TRPV channels21,22,24,30–32. In fact, the capsaicin-bound structure of truncated rTRPV1 is dilated
α-to-π transition in S6, which results in ~100° rotation of the compared to its apo-state structure6–8 (Supplementary Fig. 2f).
bottom portion of S6 and exposure of a completely different set of Whether the difference in the pore size and the presence of the α-
residues to the channel pore, has been shown experimentally by to-π transition in S6 originates from usage of different TRPV1
determining structures of TRPV6 (ref. 33), TRPV3 (refs. 21,22) and orthologs or from truncated versus full-length constructs of the
TRPV2 (ref. 34) in different conformations. By solving structures channel remains to be determined. The fact that the pore of
of sqTRPV1 in the apo, RTX-bound and capsaicin-bound states, sqTRPV1 or rTRPV1 (refs. 6–8) remains closed, while the agonists
we now demonstrate experimentally that the α-to-π transition in RTX or capsaicin are bound to the channel suggests that the

NATURE COMMUNICATIONS | (2021)12:2154 | https://doi.org/10.1038/s41467-021-22507-3 | www.nature.com/naturecommunications 5


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-021-22507-3

a b c d
50
M646 M646
M646 M646 M646 M646
P P Selectivity
P P S6 P G645 P S6 G645 Filter

Distance Along Pore (Å)


S6 G645 I644 40
I644 V669 I644 V669 S6
V669 S6 S6 T643
T643 T643 T672 T672 Central
Y673 Y673 T672
T672
S S L677
T672 T672
S S L677 Y673 D D Y673 30 cavity
L676 L676
L676 N678 L676 N678
L676 L677 L677
L680 I681 L680 I681 L680 L676
L680 N678 N678
I681 L680 I681
M684 M684 M684 M684 A682 20 Gate
A682 A682 A682
V688 V688 M684 M684
T687 T687 T687 T687 T687 T687
N689 N689
V688 10
V688
N689

Apo RTX Capsaicin 0


0 1 2 3 4 5 6 7
Pore Radius (Å)

Fig. 4 Ion channel pore. a–c Pore-forming domains in the apo (a), RTX-bound (b) and capsaicin-bound (c) states with the residues lining the pore shown
as sticks. Only two of four subunits are shown, with the front and back subunits omitted for clarity. The pore profiles are shown as space-filling models
(gray). The region that undergoes α-to-π transition in S6 is highlighted in pink. d The pore radius for the sqTRPV1 structures calculated using HOLE47. The
vertical dashed line denotes the radius of a water molecule, 1.4 Å.

channel resides in inactivated states. Future studies are required Cryo-EM sample preparation and data collection. Au/Au grids were prepared as
to better understand the role of these conformations in the gating described in the literature36. Briefly, grids were prepared by first coating C-flat
(Protochips, Inc., Morrisville, NC) CF-1.2/1.3–2Au mesh holey carbon grids with
mechanism of TRPV1. ~60 nm gold, using an Edwards Auto 306 evaporator. Subsequently, an Ar/O2
plasma treatment (4 min, 50 watts, 35.0 sccm Ar, 11.5 sccm O2) was used to remove
the carbon with a Gatan Solarus (model 950) Advanced Plasma Cleaning System
Methods (Gatan, Pleasanton, CA, USA). The grids were again plasma treated (H2/O2, 25 s,
Construct. The full-length 13-lined ground squirrel TRPV1 (GenBank 10 watts, 6.4 sccm H2, 27.5 sccm O2) prior to sample application to make their
KU877439.1, residues 1–840)5 used in cryo-EM studies was cloned into a pEG surfaces hydrophilic.
BacMam vector35, followed by thrombin cleavage site (LVPRG), enhanced green RTX and capsaicin were dissolved in dimethyl sulfoxide (DMSO) to a
fluorescent protein (eGFP) for monitoring during expression and a C-terminal concentration of 10 mM and 50 mM, respectively, and further diluted in buffer
streptavidin affinity tag (WSHPQFEK) for purification. containing 20 mM Tris (pH 8.0), 150 mM NaCl, 1 mM βME and 0.01% GDN to a
concentration of 1.5 mM. Ligands were added to protein 30 min before grid
preparation at the final concentration of 50 μM (RTX) and 300 μM (capsaicin).
Expression and purification. sqTRPV1 construct was expressed and purified as
Images of frozen-hydrated particles of sqTRPV1 (3.8 mg ml−1) in apo state
previously described for mTRPV3 (refs. 22,23), with minor modifications. Bacmids
were collected on a Titan Krios transmission electron microscope (TEM; Thermo
and baculoviruses were produced using a standard method35. Briefly, baculovirus
Fisher Scientific) operating at 300 kV, and equipped with a post-column GIF
was made in Sf9 cells for ~72 h (Thermo Fisher Scientific, mycoplasma test
Quantum energy filter and a Gatan K3 Summit direct electron detection (DED)
negative, GIBCO #12659017) and was added to the suspension adapted HEK 293
camera (Gatan, Pleasanton, CA, USA) using Leginon. A total number of 11,191
cells lacking N-acetyl-glucosaminyltransferase I (GnTI−, mycoplasma test negative,
micrographs were collected in counting mode with an image pixel size of 1.06 Å
ATCC #CRL-3022) that were maintained in Freestyle 293 media (Gibco-Life
and a defocus range of −0.8 to −2.5 µm, and energy filter slit of 30 eV. The total
Technologies #12338-018) supplemented with 2% FBS at 37 °C and 5% CO2.
dose of ~65 e− Å−2 was attained by using a dose rate of ~30 e− pixel−1 s−1 across
Twenty-four hours after transduction, 10 mM sodium butyrate was added to
50 frames for 2.5 s total exposure time.
enhance protein expression, and the temperature was reduced to 30 °C. Seventy-
Images for frozen-hydrated particles of sqTRPV1 (2.4 mg ml−1) in the presence
two hours after transduction, the cells were harvested by centrifugation at 5471 × g
of capsaicin were collected on a Titan Krios TEM (Thermo Fisher Scientific)
for 15 min using a Sorvall Evolution RC centrifuge (Thermo Fisher Scientific),
operating at 300 kV, and equipped with a post-column GIF Quantum energy filter
washed in phosphate-buffered saline (pH 8.0), and pelleted by centrifugation at
and a Gatan K3 Summit DED camera (Gatan, Pleasanton, CA, USA) using
3202 × g for 10 min using an Eppendorf 5810 centrifuge. The cell pellet was
Leginon. A total number of 9453 micrographs were collected in counting mode
resuspended in ice-cold lysis buffer, containing 20 mM Tris (pH 8.0), 150 mM
with an image pixel size of 0.83 Å and a defocus range of −1.0 to −2.5 µm. The
NaCl, 0.8 μM aprotinin, 4.3 μM leupeptin, 2 μM pepstatin A, 1 mM phe-
total dose of ~58.5 e− Å−2 was attained by using a dose rate of ~16 e− pixel−1 s−1
nylmethylsulfonyl fluoride and 1 mM β-mercaptoethanol (βME). Cells were sub-
across 50 frames for 2.5 s total exposure time.
sequently lysed using a Misonix Sonicator with a preset programme (six cycles of
Images of frozen-hydrated particles of sqTRPV1 in the presence of RTX were
15 s “on” at the amplitude of 8 followed by 15 s “off”; this programme was repeated
collected on a Polara 300 kV TEM (FEI) with a Schottky field emission gun,
three times for optimal cell lysis) under constant stirring on ice. Unbroken cells and
cartridge loading system and a Gatan K3 Summit DED camera (Gatan, Pleasanton,
cell debris were pelleted using an Eppendorf 5810 centrifuge at 3202 × g and 4 °C
CA, USA) using Leginon. A total number of 3253 micrographs were collected in
for 10 min. The supernatant was subjected to ultracentrifugation in a Beckman
counting mode with an image pixel size of 0.95 Å and a defocus range of −1.5
Coulter ultracentrifuge using a Beckman Coulter Type 45Ti rotor at 186,000 × g
to −3.5 µm. The total dose of ~70.8 e− Å−2 was attained by using a dose rate of
and 4 °C for 1 h to pellet the membranes. The membrane pellet was mechanically
~16 e− pixel−1 s−1 across 40 frames for 4 s of total exposure time.
homogenized and solubilized in the lysis buffer supplemented with 20 mM n-
Dodecyl β-D-maltoside under stirring at 4 °C for 2 h. Insoluble material was
removed by ultracentrifugation for 40 min in a Beckman Coulter Type 45Ti rotor
at 186,000 × g, and the supernatant was added to strep resin and rotated for 14–16 Image processing. All processing was completed in RELION37 and/or
h at 4 °C. Next, the resin was washed with ten column volumes of buffer containing cryoSPARC38 (Supplementary Table 1). The initial drift and beam-induced
20 mM Tris (pH 8.0), 150 mM NaCl, 1 mM βME and 0.01% (w/v) glyco-diosgenin motions were corrected using MotionCor2 (ref. 39) algorithm implemented in
(GDN), and the protein was eluted with the same buffer supplemented with RELION, and contrast transfer function (CTF) estimation was performed using
2.5 mM D-desthiobiotin. The eluted protein was concentrated using a 100 kDa Gctf40. Following CTF estimation, micrographs were manually inspected and those
NMWL centrifugal filter (MilliporeSigma™ Amicon™) to 0.5 mg ml−1. The sample with outliers in defocus values, ice thickness and astigmatism, as well as micro-
was then digested with thrombin (1:200 mass ratio of thrombin to eluted protein) graphs with lower predicted CTF-correlated resolution were excluded from the rest
for 1 h at 22 °C, and centrifuged in a Sorvall MTX 150 Micro-Ultracentrifuge of the processing pipeline (individually assessed for each parameter relative to
(Thermo Fisher Scientific) using a S100AT4 rotor for 30 min at 66,000 × g and 4 °C overall distribution; no set threshold). Initial set of particles was manually picked in
before injecting into a size-exclusion chromatography (SEC) column. The protein RELION and further classified into 100 2D classes to create templates for the
was further purified using a Superose™ 6 10/300 GL SEC column attached to an template-based picking in RELION that were used for the final round of picking.
AKTA FPLC (GE Healthcare) and equilibrated in 150 mM NaCl, 20 mM Tris, Picked particles were further 2D- and 3D-classified in iterative classification using
1 mM βME and 0.01% GDN (pH 8.0). The tetrameric peak fractions were pooled RELION and/or cryoSPARC. The reported resolutions and local resolution pre-
and concentrated using 100 kDa NMWL centrifugal filter (MilliporeSigma™ Ami- dictions of the final maps were calculated in RELION and cryoSPARC, with the
con™) to 2.4–3.8 mg ml−1. resolution range estimated by the gold standard Fourier shell correlation 0.143

6 NATURE COMMUNICATIONS | (2021)12:2154 | https://doi.org/10.1038/s41467-021-22507-3 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-021-22507-3 ARTICLE

criterion41. EM density visualization was done in UCSF Chimera42 and UCSF 6. Cao, E., Liao, M., Cheng, Y. & Julius, D. TRPV1 structures in distinct
ChimeraX43. conformations reveal activation mechanisms. Nature 504, 113–118 (2013).
7. Gao, Y., Cao, E., Julius, D. & Cheng, Y. TRPV1 structures in nanodiscs reveal
Model building. To build models of TRPV1 in Coot44, we used the previously mechanisms of ligand and lipid action. Nature 534, 347–351 (2016).
published cryo-EM structures of TRPV1 as guides. The models were tested for 8. Liao, M., Cao, E., Julius, D. & Cheng, Y. Structure of the TRPV1 ion channel
overfitting by shifting their coordinates by 0.5 Å (using shake) in Phenix, refining determined by electron cryo-microscopy. Nature 504, 107–112 (2013).
each shaken model against a corresponding unfiltered half map, and generating 9. Vlachová, V. et al. Functional role of C-terminal cytoplasmic tail of rat
densities from the resulting models in Chimera. Structures were visualized, and vanilloid receptor 1. J. Neurosci. 23, 1340–1350 (2003).
figures were prepared in UCSF Chimera, UCSF ChimeraX and Pymol45. 10. Brauchi, S., Orta, G., Salazar, M., Rosenmann, E. & Latorre, R. A hot-sensing
cold receptor: C-terminal domain determines thermosensation in transient
receptor potential channels. J. Neurosci. 26, 4835–4840 (2006).
Patch-clamp electrophysiology. sqTRPV1-pMO5 or sqTRPV1 d606–628-pMO
11. Brauchi, S. et al. Dissection of the components for PIP2 activation and
constructs were co-transfected with pcDNA3-GFP into HEK293TΔPIEZO1 (ref. 46)
thermosensation in TRP channels. Proc. Natl Acad. Sci. USA 104,
using Lipofectamine 3000 (Thermo Fisher), according to the manufacturer’s
10246–10251 (2007).
instructions. The following day, cells were plated onto glass coverslips coated with
12. Grandl, J. et al. Temperature-induced opening of TRPV1 ion channel is
Matrigel Matrix (BD Bioscience) and analysed by voltage-clamp electrophysiology
within 24 h after plating. Bath solution contained (in mM): 140 KCl, 10 HEPES, stabilized by the pore domain. Nat. Neurosci. 13, 708–714 (2010).
1 MgCl2, 10 glucose, pH 7.3 (pH adjusted with KOH) and the pipette solution 13. Gracheva, E. O. et al. Ganglion-specific splicing of TRPV1 underlies infrared
contained (in mM): 140 NaCl, 5 KCl, 10 HEPES, 1 EGTA, 1 MgCl2, pH 7.4 sensation in vampire bats. Nature 476, 88–91 (2011).
(pH adjusted with NaOH). To form the gigaohm seal, positive (+10 mmHg) 14. Prescott, E. D. & Julius, D. A modular PIP2 binding site as a determinant of
pressure was maintained inside the electrode while approaching the cell, and capsaicin receptor sensitivity. Science 300, 1284–1288 (2003).
released upon touching the cell membrane. Spontaneous single-channel events 15. Yang, F., Cui, Y., Wang, K. & Zheng, J. Thermosensitive TRP channel pore
were recorded in the cell-attached mode at room temperature at indicated voltages. turret is part of the temperature activation pathway. Proc. Natl Acad. Sci. USA
Single-channel openings are shown as upward deflections, representing outward 107, 7083–7088 (2010).
current. Currents were sampled at 25 kHz and filtered at 1 kHz. Single-channel 16. Yao, J., Liu, B. & Qin, F. Modular thermal sensors in temperature-gated
data was analysed using Clampfit 10.8. Mean single-channel amplitudes were transient receptor potential (TRP) channels. Proc. Natl Acad. Sci. USA 108,
calculated by fitting the current-amplitude histograms with Gaussian curves. The 11109–11114 (2011).
unitary conductance for sqTRPV1 was calculated by fitting the slope of the 17. Zhang, F. et al. Heat activation is intrinsic to the pore domain of TRPV1. Proc.
current–voltage relationship to the linear equation. NPo and mean open time Natl Acad. Sci. USA 115, E317–E324 (2018).
values were calculated from 10 s recordings using Clampfit. RTX (Alomone, 2 mM 18. Clapham, D. E. & Miller, C. A thermodynamic framework for understanding
stock in ethanol) was applied by perfusion. temperature sensing by transient receptor potential (TRP) channels. Proc. Natl
Acad. Sci. USA 108, 19492–19497 (2011).
Oocyte electrophysiology. Defolliculated Xenopus laevis oocytes, stages V–VI 19. Pumroy, R. A. et al. Molecular mechanism of TRPV2 channel modulation by
(Xenopus 1), were injected with sqTRPV1 mRNA (10 ng) and assayed 48–72 h cannabidiol. Elife 8, e48792 (2019).
later by two-electrode voltage clamp in calcium-free ND96, containing (in mM): 20. Zubcevic, L., Hsu, A. L., Borgnia, M. J. & Lee, S. Y. Symmetry transitions during
3 KCl, 96 NaCl, 2 MgCl2 and 10 HEPES pH 7.4 (with NaOH), supplemented with gating of the TRPV2 ion channel in lipid membranes. Elife 8, e45779 (2019).
200 nM or 2 µM capsaicin. Currents were evoked by 200-ms voltage steps from 21. Zubcevic, L. et al. Conformational ensemble of the human TRPV3 ion
−100 to 50 mV, in 10 mV increments, from a holding potential of 0 mV, and channel. Nat. Commun. 9, 4773 (2018).
digitized at 2 kHz. The reversal potential was estimated from current–voltage 22. Singh, A. K., McGoldrick, L. L. & Sobolevsky, A. I. Structure and gating
plots. Capsaicin (Sigma-Aldrich, 1 mM stock in DMSO) was applied by perfusion. mechanism of the transient receptor potential channel TRPV3. Nat. Struct.
For temperature sensitivity experiments, currents were recorded in a gap-free Mol. Biol. 25, 805–813 (2018).
mode at a holding potential of −80 mV. Temperature was controlled using the 23. Singh, A. K. et al. Structural basis of temperature sensation by the TRP
SC-20 in-line heater (Warner) and monitored by a thermistor placed in the channel TRPV3. Nat. Struct. Mol. Biol. 26, 994–998 (2019).
chamber near the oocyte. 24. Saotome, K., Singh, A. K., Yelshanskaya, M. V. & Sobolevsky, A. I. Crystal
structure of the epithelial calcium channel TRPV6. Nature 534, 506–511
(2016).
Reporting summary. Further information on research design is available in the Nature
25. Hughes, T. E. T. et al. Structural basis of TRPV5 channel inhibition by
Research Reporting Summary linked to this article.
econazole revealed by cryo-EM. Nat. Struct. Mol. Biol. 25, 53–60 (2018).
26. Yang, F. et al. An unorthodox mechanism underlying voltage sensitivity of
Data availability TRPV1 ion channel. Adv. Sci. 7, 2000575 (2020).
All data needed to evaluate the conclusions of the paper are present in the paper and/or 27. Elokely, K. et al. Understanding TRPV1 activation by ligands: Insights from
the Supplementary Materials. Cryo-EM density maps have been deposited to the Electron the binding modes of capsaicin and resiniferatoxin. Proc. Natl Acad. Sci. USA
Microscopy Data Bank (EMDB) under the accession codes EMD-23491 (sqTRPV1 apo), 113, E137–E145 (2016).
EMD-23492 (sqTRPV1 RTX), and EMD-23493 (sqTRPV1 capsaicin; see Supplementary 28. Vu, S., Singh, V., Wulff, H., Yarov-Yarovoy, V. & Zheng, J. New capsaicin
Table 1). The corresponding model coordinates have been deposited to the PDB under analogs as molecular rulers to define the permissive conformation of the
accession numbers 7LQY (sqTRPV1 apo), 7LQZ (sqTRPV1 RTX), and 7LR0 (sqTRPV1 mouse TRPV1 ligand-binding pocket. Elife 9, e62039 (2020).
capsaicin; see Supplementary Table 1). All other data are available from the 29. Kasimova, M. A. et al. A hypothetical molecular mechanism for TRPV1
corresponding author upon request. Source data are provided with this paper. activation that invokes rotation of an S6 asparagine. J. Gen. Physiol. 150,
1554–1566 (2018).
30. Zubcevic, L. et al. Cryo-electron microscopy structure of the TRPV2 ion
Received: 7 January 2021; Accepted: 18 March 2021; channel. Nat. Struct. Mol. Biol. 23, 180–186 (2016).
31. Huynh, K. W. et al. Structure of the full-length TRPV2 channel by cryo-EM.
Nat. Commun. 7, 11130 (2016).
32. Deng, Z. et al. Cryo-EM and X-ray structures of TRPV4 reveal insight into ion
permeation and gating mechanisms. Nat. Struct. Mol. Biol. 25, 252–260
(2018).
References 33. McGoldrick, L. L. et al. Opening of the human epithelial calcium channel
1. Hoffstaetter, L. J., Bagriantsev, S. N. & Gracheva, E. O. TRPs et al.: a molecular TRPV6. Nature 553, 233–237 (2018).
toolkit for thermosensory adaptations. Pflug. Arch. 470, 745–759 (2018). 34. Dosey, T. L. et al. Structures of TRPV2 in distinct conformations provide
2. Caterina, M. J. et al. The capsaicin receptor: a heat-activated ion channel in the insight into role of the pore turret. Nat. Struct. Mol. Biol. 26, 40–49 (2019).
pain pathway. Nature 389, 816–824 (1997). 35. Goehring, A. et al. Screening and large-scale expression of membrane proteins
3. Caterina, M. J. et al. Impaired nociception and pain sensation in mice lacking in mammalian cells for structural studies. Nat. Protoc. 9, 2574–2585 (2014).
the capsaicin receptor. Science 288, 306–313 (2000). 36. Russo, C. J. & Passmore, L. A. Electron microscopy: ultrastable gold substrates
4. Vandewauw, I. et al. A TRP channel trio mediates acute noxious heat sensing. for electron cryomicroscopy. Science 346, 1377–1380 (2014).
Nature 555, 662–666 (2018). 37. Scheres, S. H. RELION: implementation of a Bayesian approach to cryo-EM
5. Laursen, W. J., Schneider, E. R., Merriman, D. K., Bagriantsev, S. N. & structure determination. J. Struct. Biol. 180, 519–530 (2012).
Gracheva, E. O. Low-cost functional plasticity of TRPV1 supports heat 38. Punjani, A., Rubinstein, J. L., Fleet, D. J. & Brubaker, M. A. cryoSPARC:
tolerance in squirrels and camels. Proc. Natl Acad. Sci. USA 113, 11342–11347 algorithms for rapid unsupervised cryo-EM structure determination. Nat.
(2016). Methods 14, 290–296 (2017).

NATURE COMMUNICATIONS | (2021)12:2154 | https://doi.org/10.1038/s41467-021-22507-3 | www.nature.com/naturecommunications 7


ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-021-22507-3

39. Zheng, S. Q. et al. MotionCor2: anisotropic correction of beam-induced K.D.N., A.N. and A.I.S. analysed structural data. Y.A.N. performed single-channel and
motion for improved cryo-electron microscopy. Nat. Methods 14, 331–332 inside-out patch-clamp electrophysiology. L.A.M. performed two-electrode voltage-
(2017). clamp recordings. K.D.N., A.N., E.O.G, S.N.B. and A.I.S. wrote the manuscript.
40. Zhang, K. Gctf: real-time CTF determination and correction. J. Struct. Biol.
193, 1–12 (2016).
41. Henderson, R. et al. Outcome of the first electron microscopy validation task
Competing interests
The authors declare no competing interests.
force meeting. Structure 20, 205–214 (2012).
42. Pettersen, E. F. et al. UCSF Chimera–a visualization system for exploratory
research and analysis. J. Comput. Chem. 25, 1605–1612 (2004). Additional information
43. Pettersen, E. F. et al. UCSF ChimeraX: structure visualization for researchers, Supplementary information The online version contains supplementary material
educators, and developers. Protein Sci. 30, 70–82 (2021). available at https://doi.org/10.1038/s41467-021-22507-3.
44. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics.
Acta Crystallogr. D Biol. Crystallogr. 60, 2126–2132 (2004). Correspondence and requests for materials should be addressed to A.I.S.
45. DeLano, W. L. The PyMOL Molecular Graphics System (DeLano Scientific,
2002). Peer review information Nature Communications thanks Makoto Tominaga, Jie Zheng,
46. Lukacs, V. et al. Impaired PIEZO1 function in patients with a novel autosomal and other, anonymous, reviewers for their contributions to the peer review of this work.
recessive congenital lymphatic dysplasia. Nat. Commun. 6, 8329 (2015).
47. Smart, O. S., Neduvelil, J. G., Wang, X., Wallace, B. A. & Samsom, M. S. Reprints and permission information is available at http://www.nature.com/reprints
HOLE: a program for the analysis of the pore dimensions of ion channel
structural models. J. Mol. Graph. 14, 354–360 (1996). Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional affiliations.

Acknowledgements
Some of this work was performed at the Columbia University Cryo-Electron Microscopy Open Access This article is licensed under a Creative Commons
Center. The work was also performed at the National Center for CryoEM Access and Attribution 4.0 International License, which permits use, sharing,
Training (NCCAT) and the Simons Electron Microscopy Center located at the New York adaptation, distribution and reproduction in any medium or format, as long as you give
Structural Biology Center, supported by the NIH Common Fund Transformative High- appropriate credit to the original author(s) and the source, provide a link to the Creative
Resolution Cryo-Electron Microscopy programme (U24 GM129539), and by grants from Commons license, and indicate if changes were made. The images or other third party
the Simons Foundation (SF349247) and NY State Assembly. E.O.G. was supported by material in this article are included in the article’s Creative Commons license, unless
grants from NIH (1R01NS091300) and NSF (1754286). S.N.B. was supported by grants indicated otherwise in a credit line to the material. If material is not included in the
from NIH (1R01NS097547) and NSF (1923127). A.I.S. was supported by the NIH (R01 article’s Creative Commons license and your intended use is not permitted by statutory
CA206573, R01 NS083660, R01 NS107253) and NSF (1818086).
regulation or exceeds the permitted use, you will need to obtain permission directly from
the copyright holder. To view a copy of this license, visit http://creativecommons.org/
licenses/by/4.0/.
Author contributions
K.D.N. and A.N. made constructs, prepared protein samples, developed and optimized
cryo-EM grid preparation, and carried out cryo-EM data collection and processing. © The Author(s) 2021

8 NATURE COMMUNICATIONS | (2021)12:2154 | https://doi.org/10.1038/s41467-021-22507-3 | www.nature.com/naturecommunications

You might also like